MicroRNAs and Oxidative Stress: Comparison
Please note this is a comparison between Version 2 by Karina Chen and Version 1 by Víctor M. Víctor.

Emerging evidence reveals that oxidative stress has a critical role in the development of type 2 diabetes. Overproduction of reactive oxygen species can promote an imbalance between the production and neutralization of antioxidant defence systems, thus favoring lipid accumulation, cellular stress, and the activation of cytosolic signaling pathways, and inducing β-cell dysfunction, insulin resistance, and tissue inflammation. Over the last few years, microRNAs (miRNAs) have attracted growing attention as important mediators of diverse aspects of oxidative stress. These small endogenous non-coding RNAs of 19–24 nucleotides act as negative regulators of gene expression, including the modulation of redox signaling pathways. 

  • microRNA
  • oxidative stress
  • redox signaling
  • type 2 diabetes
Please wait, diff process is still running!

References

  1. Ighodaro, O.M. Molecular pathways associated with oxidative stress in diabetes mellitus. Biomed. Pharmacother. 2018, 108, 656–662.
  2. Siasos, G.; Paschou, S.A.; Tousoulis, D. Mitochondria and diabetes. Ann. Transl. Med. 2020, 8, 262.
  3. Mailloux, R.J. An update on mitochondrial reactive oxygen species production. Antioxidants 2020, 9, 472.
  4. Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-mediated cellular signaling. Oxid. Med. Cell Longev. 2016, 2016, 4350965.
  5. Rains, J.L.; Jain, S.K. Oxidative stress, insulin signaling, and diabetes. Free Radic. Biol. Med. 2011, 50, 567–575.
  6. Giacco, F.; Brownlee, M. Oxidative stress and diabetic complications. Circ. Res. 2010, 107, 1058–1070.
  7. Pitocco, D.; Tesauro, M.; Alessandro, R.; Ghirlanda, G.; Cardillo, C. Oxidative stress in diabetes: Implications for vascular and other complications. Int. J. Mol. Sci. 2013, 14, 21525–21550.
  8. Brubaker, P.L.; Drucker, D.J. Minireview: Glucagon-like peptides regulate cell proliferation and apoptosis in the pancreas, gut, and central nervous system. Endocrinology 2004, 145, 2653–2659.
  9. Goldin, A.; Beckman, J.A.; Schmidt, A.M.; Creager, M.A. Advanced glycation end products: Sparking the development of diabetic vascular injury. Circulation 2006, 114, 597–605.
  10. Bierhaus, A.; Hofmann, M.A.; Ziegler, R.; Nawroth, P.P. AGEs and their interaction with AGE-receptors in vascular disease and diabetes mellitus. I. The AGE concept. Cardiovas. Res. 1998, 37, 586–600.
  11. Yamagishi, S.; Nakamura, N.; Suematsu, M.; Kaseda, K.; Matsui, T. Advanced glycation end products: A molecular target for vascular complications in diabetes. Mol. Med. 2015, 21, S32–S40.
  12. Wautier, J.L.; Zoukourian, C.; Chappey, O.; Wautier, M.P.; Guillausseau, P.J.; Cao, R.; Hori, O.; Stern, D.; Schmidt, A.M. Receptor-mediated endothelial cell dysfunction in diabetic vasculopathy. Soluble receptor for advanced glycation end products blocks hyperpermeability in diabetic rats. J. Clin. Investig. 1996, 97, 238–243.
  13. Santilli, F.; D’Ardes, D.; Davi, G. Oxidative stress in chronic vascular disease: From prediction to prevention. Vasc. Pharmacol. 2015, 74, 23–37.
  14. Zhao, Z.; Zhao, C.; Zhang, X.H.; Zheng, F.; Cai, W.; Vlassara, H.; Ma, Z.A. Advanced glycation end products inhibit glucose-stimulated insulin secretion through nitric oxide-dependent inhibition of cytochrome c oxidase and adenosine triphosphate synthesis. Endocrinology 2009, 150, 2569–2576.
  15. Wang, X.; Yu, S.; Wang, C.Y.; Wang, Y.; Liu, H.X.; Cui, Y.; Zhang, L.D. Advanced glycation end products induce oxidative stress and mitochondrial dysfunction in SH-SY5Y cells. In Vitro Cell Dev. Biol. Anim. 2015, 51, 204–209.
  16. Davis, K.E.; Prasad, C.; Vijayagopal, P.; Juma, S.; Imrhan, V. Advanced glycation end products, inflammation, and chronic metabolic diseases: Links in a chain? Crit. Rev. Food Sci. Nutr. 2016, 56, 989–998.
  17. Li, Y.; Chang, Y.; Ye, N.; Chen, Y.; Zhang, N.; Sun, Y. Advanced glycation end products-induced mitochondrial energy metabolism dysfunction alters proliferation of human umbilical vein endothelial cells. Mol. Med. Rep. 2017, 15, 2673–2680.
  18. Kokkola, R.; Andersson, A.; Mullins, G.; Ostberg, T.; Treutiger, C.J.; Arnold, B.; Nawroth, P.; Andersson, U.; Harris, R.A.; Harris, H.E. RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand. J. Immunol. 2005, 61, 1–9.
  19. Leclerc, E.; Fritz, G.; Vetter, S.W.; Heizmann, C.W. Binding of S100 proteins to RAGE: An update. Biochim. Biophys. Acta 2009, 1793, 993–1007.
  20. de Paula, M.L.A.; Villela, A.M.R.; Negri, M.M.; Kanaan, S.; Weide, L.d.C.C. Role of advanced glycation end products related to the onset of diabetic kidney disease complications. Clin. Biomed. Res. 2017, 37.
  21. Lin, N.; Zhang, H.; Su, Q. Advanced glycation end-products induce injury to pancreatic beta cells through oxidative stress. Diabetes Metab. 2012, 38, 250–257.
  22. Schmitz-Peiffer, C.; Biden, T.J. Protein kinase C function in muscle, liver, and beta-cells and its therapeutic implications for type 2 diabetes. Diabetes 2008, 57, 1774–1783.
  23. Paneni, F.; Beckman, J.A.; Creager, M.A.; Cosentino, F. Diabetes and vascular disease: Pathophysiology, clinical consequences, and medical therapy: Part I. Eur. Heart J. 2013, 34, 2436–2443.
  24. Domingueti, C.P.; Dusse, L.M.; Carvalho, M.; de Sousa, L.P.; Gomes, K.B.; Fernandes, A.P. Diabetes mellitus: The linkage between oxidative stress, inflammation, hypercoagulability and vascular complications. J. Diabetes Complic. 2016, 30, 738–745.
  25. Schmitz-Peiffer, C. Protein kinase C and lipid-induced insulin resistance in skeletal muscle. Ann. N. Y. Acad. Sci. 2002, 967, 146–157.
  26. Cazzolli, R.; Mitchell, T.W.; Burchfield, J.G.; Pedersen, D.J.; Turner, N.; Biden, T.J.; Schmitz-Peiffer, C. Dilinoleoyl-phosphatidic acid mediates reduced IRS-1 tyrosine phosphorylation in rat skeletal muscle cells and mouse muscle. Diabetologia 2007, 50, 1732–1742.
  27. Keane, K.N.; Cruzat, V.F.; Carlessi, R.; de Bittencourt, P.I., Jr.; Newsholme, P. Molecular events linking oxidative stress and inflammation to insulin resistance and beta-cell dysfunction. Oxid. Med. Cell Longev. 2015, 2015, 181643.
  28. Newsholme, P.; Rebelato, E.; Abdulkader, F.; Krause, M.; Carpinelli, A.; Curi, R. Reactive oxygen and nitrogen species generation, antioxidant defenses, and beta-cell function: A critical role for amino acids. J. Endocrinol. 2012, 214, 11–20.
  29. Kim-Muller, J.Y.; Fan, J.; Kim, Y.J.; Lee, S.A.; Ishida, E.; Blaner, W.S.; Accili, D. Aldehyde dehydrogenase 1a3 defines a subset of failing pancreatic beta cells in diabetic mice. Nat. Commun. 2016, 7, 12631.
  30. Szkudelski, T.; Szkudelska, K. The relevance of AMP-activated protein kinase in insulin-secreting beta cells: A potential target for improving beta cell function? J. Physiol. Biochem. 2019, 75, 423–432.
  31. Zhang, Y.; Yamamoto, T.; Hisatome, I.; Li, Y.; Cheng, W.; Sun, N.; Cai, B.; Huang, T.; Zhu, Y.; Li, Z.; et al. Uric acid induces oxidative stress and growth inhibition by activating adenosine monophosphate-activated protein kinase and extracellular signal-regulated kinase signal pathways in pancreatic beta cells. Mol. Cell Endocrinol. 2013, 375, 89–96.
  32. Blandino-Rosano, M.; Chen, A.Y.; Scheys, J.O.; Alejandro, E.U.; Gould, A.P.; Taranukha, T.; Elghazi, L.; Cras-Meneur, C.; Bernal-Mizrachi, E. mTORC1 signaling and regulation of pancreatic beta-cell mass. Cell Cycle 2012, 11, 1892–1902.
  33. Chau, G.C.; Im, D.U.; Kang, T.M.; Bae, J.M.; Kim, W.; Pyo, S.; Moon, E.Y.; Um, S.H. mTOR controls ChREBP transcriptional activity and pancreatic beta cell survival under diabetic stress. J. Cell Biol. 2017, 216, 2091–2105.
  34. Maedler, K.; Ardestani, A. mTORC in beta cells: More than only recognizing comestibles. J. Cell Biol. 2017, 216, 1883–1885.
  35. Alhawiti, N.M.; Al Mahri, S.; Aziz, M.A.; Malik, S.S.; Mohammad, S. TXNIP in metabolic regulation: Physiological role and therapeutic outlook. Curr. Drug Targets 2017, 18, 1095–1103.
  36. Saxena, G.; Chen, J.; Shalev, A. Intracellular shuttling and mitochondrial function of thioredoxin-interacting protein. J. Biol. Chem. 2010, 285, 3997–4005.
  37. Sokolova, M.; Sahraoui, A.; Hoyem, M.; Ogaard, J.; Lien, E.; Aukrust, P.; Yndestad, A.; Ranheim, T.; Scholz, H. NLRP3 inflammasome mediates oxidative stress-induced pancreatic islet dysfunction. Am. J. Physiol. Endocrinol. Metab. 2018, 315, E912–E923.
  38. Zhao, Y.; Sun, H.; Li, X.; Zha, Y.; Hou, W. Hydroxysafflor yellow A attenuates high glucose-induced pancreatic beta-cells oxidative damage via inhibiting JNK/c-jun signaling pathway. Biochem. Biophys. Res. Commun. 2018, 505, 353–359.
  39. Yung, J.H.M.; Giacca, A. Role of c-Jun N-terminal kinase (JNK) in obesity and type 2 diabetes. Cells 2020, 9, 706.
  40. Hayes, H.L.; Moss, L.G.; Schisler, J.C.; Haldeman, J.M.; Zhang, Z.; Rosenberg, P.B.; Newgard, C.B.; Hohmeier, H.E. Pdx-1 activates islet alpha- and beta-cell proliferation via a mechanism regulated by transient receptor potential cation channels 3 and 6 and extracellular signal-regulated kinases 1 and 2. Mol. Cell Biol. 2013, 33, 4017–4029.
  41. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233.
  42. Miyoshi, K.; Miyoshi, T.; Siomi, H. Many ways to generate microRNA-like small RNAs: Non-canonical pathways for microRNA production. Mol. Genet. Genom. 2010, 284, 95–103.
  43. Catalanotto, C.; Cogoni, C.; Zardo, G. MicroRNA in control of gene expression: An overview of nuclear functions. Int. J. Mol. Sci. 2016, 17, 1712.
  44. Eliasson, L.; Esguerra, J.L. Role of non-coding RNAs in pancreatic beta-cell development and physiology. Acta Physiol. 2014, 211, 273–284.
  45. Chen, H.; Lan, H.Y.; Roukos, D.H.; Cho, W.C. Application of microRNAs in diabetes mellitus. J. Endocrinol. 2014, 222, R1–R10.
  46. Golbidi, S.; Ebadi, S.A.; Laher, I. Antioxidants in the treatment of diabetes. Curr. Diabetes Rev. 2011, 7, 106–125.
  47. La Sala, L.; Mrakic-Sposta, S.; Tagliabue, E.; Prattichizzo, F.; Micheloni, S.; Sangalli, E.; Specchia, C.; Uccellatore, A.C.; Lupini, S.; Spinetti, G.; et al. Circulating microRNA-21 is an early predictor of ROS-mediated damage in subjects with high risk of developing diabetes and in drug-naive T2D. Cardiovasc. Diabetol. 2019, 18, 18.
  48. La Sala, L.; Mrakic-Sposta, S.; Micheloni, S.; Prattichizzo, F.; Ceriello, A. Glucose-sensing microRNA-21 disrupts ROS homeostasis and impairs antioxidant responses in cellular glucose variability. Cardiovasc. Diabetol. 2018, 17, 105.
  49. Gao, L.; Liu, Y.; Guo, S.; Xiao, L.; Wu, L.; Wang, Z.; Liang, C.; Yao, R.; Zhang, Y. LAZ3 protects cardiac remodeling in diabetic cardiomyopathy via regulating miR-21/PPARa signaling. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 3322–3338.
  50. Dubois, V.; Eeckhoute, J.; Lefebvre, P.; Staels, B. Distinct but complementary contributions of PPAR isotypes to energy homeostasis. J. Clin. Investig. 2017, 127, 1202–1214.
  51. Magenta, A.; Cencioni, C.; Fasanaro, P.; Zaccagnini, G.; Greco, S.; Sarra-Ferraris, G.; Antonini, A.; Martelli, F.; Capogrossi, M.C. miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition. Cell Death Differ. 2011, 18, 1628–1639.
  52. Belgardt, B.F.; Ahmed, K.; Spranger, M.; Latreille, M.; Denzler, R.; Kondratiuk, N.; von Meyenn, F.; Villena, F.N.; Herrmanns, K.; Bosco, D.; et al. The microRNA-200 family regulates pancreatic beta cell survival in type 2 diabetes. Nat. Med. 2015, 21, 619–627.
  53. Carlomosti, F.; D’Agostino, M.; Beji, S.; Torcinaro, A.; Rizzi, R.; Zaccagnini, G.; Maimone, B.; Di Stefano, V.; De Santa, F.; Cordisco, S.; et al. Oxidative stress-induced miR-200c disrupts the regulatory loop among SIRT1, FOXO1, and eNOS. Antioxid. Redox Signal. 2017, 27, 328–344.
  54. Yamakuchi, M. MicroRNA Regulation of SIRT1. Front. Physiol. 2012, 3, 68.
  55. Li, Q.; Kim, Y.R.; Vikram, A.; Kumar, S.; Kassan, M.; Gabani, M.; Lee, S.K.; Jacobs, J.S.; Irani, K. P66Shc-induced microRNA-34a causes diabetic endothelial dysfunction by downregulating Sirtuin1. Arter. Thromb. Vasc. Biol. 2016, 36, 2394–2403.
  56. Kassan, M.; Vikram, A.; Li, Q.; Kim, Y.R.; Kumar, S.; Gabani, M.; Liu, J.; Jacobs, J.S.; Irani, K. MicroRNA-204 promotes vascular endoplasmic reticulum stress and endothelial dysfunction by targeting Sirtuin1. Sci. Rep. 2017, 7, 9308.
  57. Chen, D.L.; Yang, K.Y. Berberine Alleviates oxidative stress in islets of diabetic mice by inhibiting miR-106b expression and up-regulating SIRT1. J. Cell Biochem. 2017, 118, 4349–4357.
  58. Li, L.M.; Hou, D.X.; Guo, Y.L.; Yang, J.W.; Liu, Y.; Zhang, C.Y.; Zen, K. Role of microRNA-214-targeting phosphatase and tensin homolog in advanced glycation end product-induced apoptosis delay in monocytes. J. Immunol. 2011, 186, 2552–2560.
  59. Comer, F.I.; Parent, C.A. PI 3-kinases and PTEN: How opposites chemoattract. Cell 2002, 109, 541–544.
  60. Muratsu-Ikeda, S.; Nangaku, M.; Ikeda, Y.; Tanaka, T.; Wada, T.; Inagi, R. Downregulation of miR-205 modulates cell susceptibility to oxidative and endoplasmic reticulum stresses in renal tubular cells. PLoS ONE 2012, 7, e41462.
  61. Fleissner, F.; Jazbutyte, V.; Fiedler, J.; Gupta, S.K.; Yin, X.; Xu, Q.; Galuppo, P.; Kneitz, S.; Mayr, M.; Ertl, G.; et al. Short communication: Asymmetric dimethylarginine impairs angiogenic progenitor cell function in patients with coronary artery disease through a microRNA-21-dependent mechanism. Circ. Res. 2010, 107, 138–143.
  62. Wu, X.D.; Liu, W.L.; Zeng, K.; Lei, H.Y.; Zhang, Q.G.; Zhou, S.Q.; Xu, S.Y. Advanced glycation end products activate the miRNA/RhoA/ROCK2 pathway in endothelial cells. Microcirculation 2014, 21, 178–186.
  63. Poy, M.N.; Eliasson, L.; Krutzfeldt, J.; Kuwajima, S.; Ma, X.; Macdonald, P.E.; Pfeffer, S.; Tuschl, T.; Rajewsky, N.; Rorsman, P.; et al. A pancreatic islet-specific microRNA regulates insulin secretion. Nature 2004, 432, 226–230.
  64. Poy, M.N.; Hausser, J.; Trajkovski, M.; Braun, M.; Collins, S.; Rorsman, P.; Zavolan, M.; Stoffel, M. miR-375 maintains normal pancreatic alpha- and beta-cell mass. Proc. Natl. Acad. Sci. USA 2009, 106, 5813–5818.
  65. Melkman-Zehavi, T.; Oren, R.; Kredo-Russo, S.; Shapira, T.; Mandelbaum, A.D.; Rivkin, N.; Nir, T.; Lennox, K.A.; Behlke, M.A.; Dor, Y.; et al. miRNAs control insulin content in pancreatic beta-cells via downregulation of transcriptional repressors. EMBO J. 2011, 30, 835–845.
  66. Iguchi, H.; Urashima, Y.; Inagaki, Y.; Ikeda, Y.; Okamura, M.; Tanaka, T.; Uchida, A.; Yamamoto, T.T.; Kodama, T.; Sakai, J. SOX6 suppresses cyclin D1 promoter activity by interacting with beta-catenin and histone deacetylase 1, and its down-regulation induces pancreatic beta-cell proliferation. J. Biol. Chem. 2007, 282, 19052–19061.
  67. Zhao, X.; Mohan, R.; Ozcan, S.; Tang, X. MicroRNA-30d induces insulin transcription factor MafA and insulin production by targeting mitogen-activated protein 4 kinase 4 (MAP4K4) in pancreatic beta-cells. J. Biol. Chem. 2012, 287, 31155–31164.
  68. Matsuoka, T.A.; Kaneto, H.; Kawashima, S.; Miyatsuka, T.; Tochino, Y.; Yoshikawa, A.; Imagawa, A.; Miyazaki, J.; Gannon, M.; Stein, R.; et al. Preserving Mafa expression in diabetic islet beta-cells improves glycemic control in vivo. J. Biol. Chem. 2015, 290, 7647–7657.
  69. Latreille, M.; Hausser, J.; Stutzer, I.; Zhang, Q.; Hastoy, B.; Gargani, S.; Kerr-Conte, J.; Pattou, F.; Zavolan, M.; Esguerra, J.L.; et al. MicroRNA-7a regulates pancreatic beta cell function. J. Clin. Investig. 2014, 124, 2722–2735.
  70. Saxton, R.A.; Sabatini, D.M. mTOR signaling in growth, metabolism, and disease. Cell 2017, 168, 960–976.
  71. Wang, Y.; Liu, J.; Liu, C.; Naji, A.; Stoffers, D.A. MicroRNA-7 regulates the mTOR pathway and proliferation in adult pancreatic beta-cells. Diabetes 2013, 62, 887–895.
  72. Plaisance, V.; Abderrahmani, A.; Perret-Menoud, V.; Jacquemin, P.; Lemaigre, F.; Regazzi, R. MicroRNA-9 controls the expression of Granuphilin/Slp4 and the secretory response of insulin-producing cells. J. Biol. Chem. 2006, 281, 26932–26942.
  73. Burgos-Moron, E.; Abad-Jimenez, Z.; Maranon, A.M.; Iannantuoni, F.; Escribano-Lopez, I.; Lopez-Domenech, S.; Salom, C.; Jover, A.; Mora, V.; Roldan, I.; et al. Relationship between oxidative stress, ER stress, and inflammation in type 2 diabetes: The battle continues. J. Clin. Med. 2019, 8, 1385.
  74. Victor, V.M.; Rocha, M.; Herance, R.; Hernandez-Mijares, A. Oxidative stress and mitochondrial dysfunction in type 2 diabetes. Curr. Pharm. Des. 2011, 17, 3947–3958.
  75. Duarte, F.V.; Palmeira, C.M.; Rolo, A.P. The emerging role of MitomiRs in the pathophysiology of human disease. Adv. Exp. Med. Biol. 2015, 888, 123–154.
  76. Bandiera, S.; Mategot, R.; Girard, M.; Demongeot, J.; Henrion-Caude, A. MitomiRs delineating the intracellular localization of microRNAs at mitochondria. Free Radic. Biol. Med. 2013, 64, 12–19.
  77. Mohamed, J.S.; Hajira, A.; Pardo, P.S.; Boriek, A.M. MicroRNA-149 inhibits PARP-2 and promotes mitochondrial biogenesis via SIRT-1/PGC-1alpha network in skeletal muscle. Diabetes 2014, 63, 1546–1559.
  78. Bienertova-Vasku, J.; Sana, J.; Slaby, O. The role of microRNAs in mitochondria in cancer. Cancer Lett. 2013, 336, 1–7.
  79. Sun, W.; Zhao, L.; Song, X.; Zhang, J.; Xing, Y.; Liu, N.; Yan, Y.; Li, Z.; Lu, Y.; Wu, J.; et al. MicroRNA-210 modulates the cellular energy metabolism shift during H2O2-induced oxidative stress by repressing ISCU in H9c2 cardiomyocytes. Cell Physiol. Biochem. 2017, 43, 383–394.
More